Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hye-Ock Jang is active.

Publication


Featured researches published by Hye-Ock Jang.


Biochimica et Biophysica Acta | 2008

Visfatin enhances ICAM-1 and VCAM-1 expression through ROS-dependent NF-κB activation in endothelial cells

Su-Ryun Kim; Yun-Hee Bae; Soo-Kyung Bae; Kyu-Sil Choi; Kwon-Ha Yoon; Tae Hyeon Koo; Hye-Ock Jang; Il Yun; Kyu-Won Kim; Young-Guen Kwon; Mi-Ae Yoo; Moon-Kyoung Bae

Visfatin has recently been identified as a novel visceral adipokine which may be involved in obesity-related vascular disorders. However, it is not known whether visfatin directly contributes to endothelial dysfunction. Here, we investigated the effect of visfatin on vascular inflammation, a key step in a variety of vascular diseases. Visfatin induced leukocyte adhesion to endothelial cells and the aortic endothelium by induction of the cell adhesion molecules, ICAM-1 and VCAM-1. Promoter analysis revealed that visfatin-mediated induction of CAMs is mainly regulated by nuclear factor-kappaB (NF-kappaB). Visfatin stimulated IkappaBalpha phosphorylation, nuclear translocation of the p65 subunit of NF-kappaB, and NF-kappaB DNA binding activity in HMECs. Furthermore, visfatin increased ROS generation, and visfatin-induced CAMs expression and NF-kappaB activation were abrogated in the presence of the direct scavenger of ROS. Taken together, our results demonstrate that visfatin is a vascular inflammatory molecule that increases expression of the inflammatory CAMs, ICAM-1 and VCAM-1, through ROS-dependent NF-kappaB activation in endothelial cells.


FEBS Letters | 2006

Hypoxic induction of human visfatin gene is directly mediated by hypoxia-inducible factor-1

Soo-Kyung Bae; Su-Ryun Kim; Jong Gab Kim; Jee Yeon Kim; Tae Hyeon Koo; Hye-Ock Jang; Il Yun; Mi-Ae Yoo; Moon-Kyoung Bae

Visfatin has been originally identified as a growth factor for early stage B cells and recently known as an adipokine. Here, we report that hypoxia induces the visfatin mRNA and protein levels in MCF7 breast cancer cells. We also demonstrate that induction of visfatin gene is regulated by hypoxia‐inducible factor‐1α (HIF‐1α). Moreover, 5′‐flanking promoter region of human visfatin gene contains two functional HIF responsive elements (HREs), activating the expression of visfatin. Mutation of these HREs in the visfatin promoter abrogates activation of a luciferase reporter gene driven by visfatin promoter under hypoxia. Taken together, our results demonstrate that visfatin is a new hypoxia‐inducible gene of which expression is stimulated through the interaction of HIF‐1 with HRE sites in its promoter region.


Archives of Pharmacal Research | 2009

Resveratrol inhibits Porphyromonas gingivalis lipopolysaccharide-induced endothelial adhesion molecule expression by suppressing NF-κB activation

Hyun-Joo Park; Seong-Kyoon Jeong; Su-Ryun Kim; Soo-Kyung Bae; Woo-Sik Kim; Seong-Deok Jin; Tae Hyeon Koo; Hye-Ock Jang; Il Yun; Kyu-Won Kim; Moon-Kyoung Bae

P. gingivalis is a major pathogen that is involved in the onset and progression of periodontal disease. This study investigated the effect of resveratrol, a naturally occurring polyphenol, on P. gingivalis LPS-accelerated vascular inflammation, a key step in the progression of periodontitis. Resveratrol significantly inhibited the P. gingivalis LPS-induced adhesion of leukocytes to endothelial cells and to the aortic endothelium by down-regulating the cell adhesion molecules, ICAM-1 and VCAM-1. Moreover, the inhibition of the P. gingivalis LPS-induced cell adhesion molecules by resveratrol was mainly mediated by nuclear factor-κB (NF-κB). Resveratrol suppressed P. gingivalis LPS-stimulated IκBα phosphorylation and nuclear translocation of the p65 subunit of NF-κB in HMECs. Overall, these findings suggest that resveratrol significantly attenuates the P. gingivalis LPS-induced monocyte adhesion to the endothelium by suppressing the expression of the NF-κB-dependent cell adhesion molecules, suggesting its therapeutic role in periodontal pathogen-induced vascular inflammation.


Endocrinology | 2012

Curcumin Down-Regulates Visfatin Expression and Inhibits Breast Cancer Cell Invasion

Su-Ryun Kim; Hyun-Joo Park; Yun-Hee Bae; Soon-Cheol Ahn; Hee-Jun Wee; Il Yun; Hye-Ock Jang; Moon-Kyoung Bae; Soo-Kyung Bae

Obesity is frequently associated with breast cancer. Such associations are possibly mediated by adipokines. Visfatin, an adipokine, has recently been shown to be related to the development and progression of breast cancer. Therefore, the down-regulation of visfatin may be a novel strategy for breast cancer therapy. Curcumin has anticancer activities by modulating multiple signaling pathways and genes. The purpose of this study was to investigate whether visfatin gene expression is affected by curcumin in human breast cancer cells and to characterize the functional role of visfatin in breast cancer. We found that the mRNA and protein levels of visfatin were down-regulated by curcumin in MDA-MB-231, MDA-MB-468, and MCF-7 breast cancer cells, along with decreased activity of constitutive nuclear factor (NF)-κB. We confirmed the repressive effect of curcumin on visfatin transcription by performing a visfatin promoter-driven reporter assay and identified two putative NF-κB-binding sites on visfatin promoter that are important for this effect. EMSA and chromatin immunoprecipitation analysis indicated the binding of p65 to the visfatin promoter, which was effectively blocked by curcumin. Enforced expression of p65 protein increased visfatin promoter activity, whereas blocking NF-κB signaling suppressed visfatin gene expression. Visfatin could enhance the invasion of MDA-MB-231 cells and also attenuate curcumin-induced inhibition of cell invasion; on the other hand, visfatin knockdown by small interfering RNA led to the reduction of cell invasion. Our data demonstrate, for the first time, that curcumin down-regulates visfatin gene expression in human breast cancer cells by a mechanism that is, at least in part, NF-κB dependent and suggest that visfatin may contribute to breast cancer cell invasion and link obesity to breast cancer development and progression.


Experimental Cell Research | 2009

Neuromedin B induces angiogenesis via activation of ERK and Akt in endothelial cells

Hyun-Joo Park; Su-Ryun Kim; Soo-Kyung Bae; Yoon Kyung Choi; Yun-Hee Bae; Eok-Cheon Kim; Woo Jean Kim; Hye-Ock Jang; Il Yun; Young-Myeong Kim; Moon-Kyoung Bae

Neuromedin B (NMB) is one of the bombesin-like peptides in mammals. Recently, bombesin-like peptides have been characterized as growth factors in highly vascularized tumors. In this study, we report that NMB potently stimulates in vivo neovascularization in a mouse Matrigel plug and the sprouting of endothelial cells ex vivo in rat aortic rings. In addition, NMB increases the migration and tube formation in human umbilical vein endothelial cells (HUVECs). Moreover, treatment of HUVECs with NMB activates the extracellular signal-regulated kinase 1/2 (ERK(1/2)), Akt, and endothelial nitric oxide synthase (eNOS) and increases the level of NO production in a dose- and time-dependent manner. Furthermore, ERK activation and angiogenic sprouting in response to NMB are significantly blocked by the MEK inhibitor. Inhibition of phosphatidylinositol 3-kinase (PI3K) suppresses the NMB-stimulated tubular formation of HUVECs, along with reduction in the phosphorylation of Akt and eNOS. Taken together, these results indicate that NMB is a novel angiogenic peptide, and its angiogenic activity is mediated by activating the MEK/ERK- and PI3K/Akt/eNOS-dependent pathways. This study suggests that NMB may play important roles in mediating a variety of pathophysiological angiogenesis.


Biochemical and Biophysical Research Communications | 2010

Angiogenic role of orexin-A via the activation of extracellular signal-regulated kinase in endothelial cells

Mi-Kyoung Kim; Hyun-Joo Park; Su-Ryun Kim; Yoon Kyung Choi; Hwa Kyoung Shin; Jae-Hoon Jeon; Hye-Ock Jang; I. I. Yun; Soo-Kyung Bae; Moon-Kyoung Bae

Orexin-A, a neuropeptide originally discovered in the hypothalamus, is found in peripheral organs, as well as in the central nervous system, and is involved in the regulation of food intake, energy homeostasis, and cardiovascular functions. In this study, we report that orexin-A induces invivo neovascularization in a mouse Matrigel plug and ex vivo sprouting of endothelial cells in rat aortic rings. We also show that orexin-A increases migration and tube formation in human umbilical vein endothelial cells (HUVECs), and this effect is mediated by orexin receptors on endothelial cells. Moreover, orexin-A activates the extracellular signal-regulated kinase 1/2 (ERK1/2) in HUVECs, which is closely linked to angiogenic responses. The inhibition of ERK activation significantly suppresses orexin-A-stimulated endothelial angiogenesis. Taken together, our results indicate that orexin-A functions as a new proangiogenic peptide and requires MEK/ERK-dependent pathway for its angiogenic actions. These results suggest orexin-A and its receptor may act as important modulators of angiogenesis under pathophysiological conditions.


Natural Product Research | 2007

Effect of extracts from safflower seeds on osteoblast differentiation and intracellular calcium ion concentration in MC3T3-E1 cells.

Hye-Ock Jang; Young-Sik Park; Jong-Hwa Lee; Jun-Bong Seo; Kyo-Il Koo; Soo-Cheol Jeong; Seong-Deok Jin; Young-Ho Lee; Hyun-Sup Eom; Il Yun

Although safflower seeds have long been used in Korea as herbal medicines, very little research has been published on the effects of safflower seed on bone formation or bone density. The study reported here therefore examined bone nodule formation, calcium uptake, alkaline phosphatase activity, and intracellular concentration of calcium ion [Ca2+]i in murine osteoblastic cells of the MC3T3-E1 line that were cultured on modified Eagles minimal essential medium alone (controls) or with addition of 0.1% crude extract of safflower seed (experimental group I) or 0.1% aqueous fraction of safflower seed (experimental group II). Fluorescence spectrometry measurement of ([Ca2+]i) showed significantly accelerated rates of osteoblast differentiation in experimental group I (3 µL of crude extract in 8 × 104 cells) and experimental group II (2 µL of aqueous fraction in 8 × 104 cells) compared to the control group.


Biochemical and Biophysical Research Communications | 2010

Thromboxane A2 increases endothelial permeability through upregulation of interleukin-8

Su-Ryun Kim; Soo-Kyung Bae; Hyun-Joo Park; Mi-Kyoung Kim; Koanhoi Kim; Shi-Young Park; Hye-Ock Jang; Il Yun; Yung-Jin Kim; Mi-Ae Yoo; Moon-Kyoung Bae

Thromboxane A(2) (TXA(2)), a major prostanoid formed from prostaglandin H(2) by thromboxane synthase, is involved in the pathogenesis of a variety of vascular diseases. In this study, we report that TXA(2) mimetic U46619 significantly increases the endothelial permeability both in vitro and in vivo. U46619 enhanced the expression and secretion of interleukin-8 (IL-8), a major inducer of vascular permeability, in endothelial cells. Promoter analysis showed that the U46619-induced expression of IL-8 was mainly regulated by nuclear factor-kappaB (NF-kappaB). U46619 induced the activation of NF-kappaB through IkappaB kinase (IKK) activation, IkappaB phosphorylation and NF-kappaB nuclear translocation. Furthermore, the inhibition of IL-8 or blockade of the IL-8 receptor attenuated the U46619-induced endothelial cell permeability by modulating the cell-cell junctions. Overall, these results suggest that U46619 promotes vascular permeability through the production of IL-8 via NF-kappaB activation in endothelial cells.


Journal of Pineal Research | 2013

Melatonin inhibits visfatin-induced inducible nitric oxide synthase expression and nitric oxide production in macrophages

Young-Soon Kang; Yong-Gyu Kang; Hyun-Joo Park; Hee-Jun Wee; Hye-Ock Jang; Moon-Kyoung Bae; Soo-Kyung Bae

Aberrant expression of inducible nitric oxide synthase (iNOS) in macrophages, which has been reported to be suppressed by melatonin, has an important contribution in the development of pathological inflammation. Visfatin, an adipokine, regulates the expression of various inflammatory factors, leading to inflammation; however, the influence of visfatin on iNOS‐driven processes in macrophages is unclear. Here, we report the assessment of the role of visfatin in the regulation of iNOS gene expression in macrophages. Our data show that the levels of iNOS protein in peritoneal macrophages as well as nitric oxide (NO) in blood plasma were significantly lower after lipopolysaccharide treatment in visfatin+/− mice than those in the WT mice. In addition, visfatin increases iNOS mRNA and protein levels in RAW 264.7 cells, along with increasing production of NO. The enhancement of iNOS expression was prevented by treating the cells with inhibitors of the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3), nuclear factor (NF)‐κB, extracellular signal–regulated kinase 1/2, and c‐Jun N‐terminal kinase pathways. Our results also show that visfatin‐induced iNOS expression and NO production were significantly inhibited by melatonin, an effect that was closely associated with a reduction in phosphorylated JAK2/STAT3 levels and with the inhibition of p65 translocation into nucleus. In conclusion, our data show, for the first time, that melatonin suppresses visfatin‐induced iNOS upregulation in macrophages by inhibiting the STAT3 and NF‐κB pathways. Moreover, our data suggest that melatonin could be therapeutically useful for attenuating the development of visfatin–iNOS axis‐associated diseases.


Biochemical and Biophysical Research Communications | 2012

Upregulation of thromboxane synthase mediates visfatin-induced interleukin-8 expression and angiogenic activity in endothelial cells.

Su-Ryun Kim; Yun-Hoa Jung; Hyun-Joo Park; Mi-Kyoung Kim; Joo-Won Jeong; Hye-Ock Jang; Il Yun; Soo-Kyung Bae; Moon-Kyoung Bae

Thromboxane synthase (TXAS) is an enzyme that catalyzes the synthesis of thromboxane A(2) (TXA(2)). Overexpression of TXAS is associated with a variety of vascular diseases. Recently, we reported that visfatin, a novel adipokine, exhibits angiogenic actions. In this study, we showed that visfatin increased mRNA and protein levels of TXAS and stimulated TXA(2) biosynthesis in vascular endothelial cells. In addition, visfatin induced the expression and secretion of interleukin-8 (IL-8), which is blocked by a TXAS inhibitor and by the transfection of siRNA specific for TXAS. Furthermore, the inhibition of TXAS activity and blockade of the IL-8 receptor attenuated visfatin-induced endothelial angiogenesis. Together, these results showed that visfatin promoted IL-8 production by upregulation of TXAS, leading to angiogenic activation in endothelial cells.

Collaboration


Dive into the Hye-Ock Jang's collaboration.

Top Co-Authors

Avatar

Il Yun

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Moon-Kyoung Bae

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Soo-Kyung Bae

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Hyun-Joo Park

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

In-Kyo Chung

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Su-Ryun Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Moon-Kyung Bae

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Mi-Kyoung Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Yun-Hee Bae

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Seong-Deok Jin

Pusan National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge